EP2131841B1 - Methods for treating acute pain - Google Patents

Methods for treating acute pain Download PDF

Info

Publication number
EP2131841B1
EP2131841B1 EP08724934A EP08724934A EP2131841B1 EP 2131841 B1 EP2131841 B1 EP 2131841B1 EP 08724934 A EP08724934 A EP 08724934A EP 08724934 A EP08724934 A EP 08724934A EP 2131841 B1 EP2131841 B1 EP 2131841B1
Authority
EP
European Patent Office
Prior art keywords
inhibitor
ibudilast
pde
pain
opioid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Not-in-force
Application number
EP08724934A
Other languages
German (de)
English (en)
French (fr)
Other versions
EP2131841A1 (en
Inventor
Kirk W. Johnson
Linda Watkins
Mark Hutchinson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Avigen Inc
University of Colorado
Original Assignee
Avigen Inc
University of Colorado
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Avigen Inc, University of Colorado filed Critical Avigen Inc
Priority to PL08724934T priority Critical patent/PL2131841T3/pl
Publication of EP2131841A1 publication Critical patent/EP2131841A1/en
Application granted granted Critical
Publication of EP2131841B1 publication Critical patent/EP2131841B1/en
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/485Morphinan derivatives, e.g. morphine, codeine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates generally to potentiation ofopioid-induced analgesia in a subject by administration of a phosphodiesterase (PDE) inhibitor.
  • PDE phosphodiesterase
  • the present invention pertains to a PDE inhibitor, such as ibudilast (3-isobutyryl-2-isopropylpyrazolo[1,5-a]pyridine; also termed AV411 herein), in combination with an opioid analgesic, for use in treating acute pain.
  • Pain is both a sensory and emotional experience, and is generally associated with tissue damage or inflammation.
  • pain is divided into two general categories - acute pain and chronic pain. Both differ in their etiology, pathophysiology, diagnosis, and most importantly, treatment.
  • Morphine is a primary analgesic used in the treatment of pain as specified by the World Health Organization (WHO).
  • WHO World Health Organization
  • Morphine belongs to a class of compounds know as 4,5-epoxymorphinans. Once the structure of morphine had been established, attempts to synthesize opioids that were more potent, longer acting and which had fewer side effects began. This gave rise to numerous other compound classes such as morphinans, benzomorphinans, arylmorphinans, 4-phenylpiperidines, 4-anilinopiperidines and 3,3-diphenylpropylamines, all of which have differing pharmacokinetic and pharmacodynamic properties.
  • non-4,5-epoxymorphinans are levorphanol (morphinan), pethidine (4-phenylpiperidine), fentanyl (4-anilinopiperidine) and methadone (3,3-diphenylpropylamine).
  • the relief of pain by all of these clinically employed opioids is relatively selective in that other sensations (e.g ., touch, vision, hearing, smelling) are typically not compromised.
  • the analgesic benefit of morphine and related opioids can be accompanied by other undesirable side effects involving the central nervous system and gastrointestinal system including drowsiness, respiratory depression, constipation, nausea and vomiting. There are significant adverse (respiratory depression) and unpleasant dose limiting side effects.
  • a characteristic feature of opioid drugs is the development of tolerance and physical dependence ( Gilman, A.G., Goodman, L.S., Rall, T.W., and Murad, F. The Pharmacological Basis of Therapeutics. New York: MacMillan Publishing Co., 1985 ).
  • opioid analgesics are ineffective in certain pain conditions (including certain neuropathic syndromes).
  • the combination of dose limiting side effects and the potential development of tolerance which requires increases in analgesic requirements results in many patients who are left without adequate pain relief by opioids.
  • Some potentiators enhanced efficacy of two agents, typically in a greater-than-additive manner
  • opioid analgesia in rodent models include ketamine ( AR Campos et al., Biol Pharm Bull, 29:86, 2006 ), dopamine antagonists ( JA Kiritsy-Roy et al., Pharmacol Biochem Behav 32:717, 1989 ), and certain non-steroidal anti-inflammatory agents ( S. Zelcer et al., Brain Res 1040:151, 2005 ).
  • ibudilast (3-isoburyryl-2-isopropylpyrazolo[1,5-a]pyridine), is a non-selective inhibitor of cyclic nucleotide phosphodiesterase (PDE) ( Fujimoto, T., et al., J. ofNeuroimmunology, 95 (1999) 35-92 ). Ibudilast also acts as an LTD4 antagonist, an anti-inflammatory, a PAF antagonist, and a vasodilatarory agent (Thompson Current Drug Reports). Ibudilast is thought to exert a neuroprotective role in the central nervous system of mammals, presumably via suppression of the activation of glial cells ( Mizuno et al.
  • Ibudilast has been widely used in Japan for relieving symptoms associated with ischemic stroke or bronchial asthma.
  • Marketed indications for ibudilast in Japan include its use as a vasodilator, for treating allergy, eye tissue regeneration, ocular disease, and treatment of allergic ophthalmic disease (Thompson Current Drug Reports).
  • Thompson Current Drug Reports In recent clinical trials, its use in the treatment of multiple selerosis, an inflammatory disease of the central nervous system, has been explored ( News.Medical.Net; Pharmaceutical News, 2 Aug 2005 ).
  • WO 2007/146087 hints at the possibility of using PDE inhibitors for the treatment of pain but does not provide data on the treatment of acute pain, nor does it provide specific data for PDE-4 inhibitors in particular.
  • ibudilast is a potent suppressor of glial activation but do not disclose the use of PDE IV inhibitors for the treatment of acute pain.
  • the invention provides a phosphodiesterase 4 inhibitor, as claimed in any of the claims as enclosed.
  • the PDE inhibitor is selected from the group consisting of Rolipram, Arofylline, Roflumilast, Pentoxyfylline, and Propenrofylline.
  • the phosphodiesterase-4 inhibitor is ibudilast.
  • the opioid is morphine or oxycodone. In certain embodiments, the PDE inhibitor is administered either prior to or concurrently with the opioid.
  • the invention provides for the above use for treating acute pain comprising administering to a subject in need thereof a therapeutically effective amount of ibudilast and an opioid.
  • the ibudilast is administered to the subject either prior to or concurrently with the opioid. If administered concurrently, the ibudilast and morphine can be in the same or different compositions.
  • the opioid is morphine or oxycodone.
  • Mammalian subjects suitable for treatment by the methods described herein include, but are not limited to, those suffering from back pain, post-operative pain, injury-related pain, or disease-related pain.
  • the subject is a human.
  • ibudilast is administered systemically, for example, via intravenous, subcutaneous, oral, intranasal, sublingual or other systemic routes. In other embodiments, ibudilast is administered centrally, for example, intrathecally. In certain embodiments, ibudilast is administered orally or intraperitoneally. In one embodiment, the opioid is administered subcutaneously.
  • a therapeutic dosage amount of ibudilast or an opioid analgesic may be achieved by intermittent administration, or administration once daily (i.e., in a single dose), twice daily (i.e., in two separate doses), three times daily, or may be administered as multiple doses over a time course of several days, weeks, or even months.
  • Such administering is typically over a duration of time effective to result in a diminution, and ideally elimination or even reversal, of acute pain.
  • Exemplary durations of treatment include at least about one week, from 1 week to 1 month, from two weeks to 2 months, up to about 6 months, up to about 12 months or even longer. In one particular embodiment, treatment lasts from about 1 week to about 50 weeks.
  • the administering is over a duration of time effective to result in elimination of acute pain.
  • the ibudilast and opioid analgesic are administered in combination with one or more other agents effective for treating pain.
  • agents include analgesics, non-steroidal anti-inflammatory drugs (NSAIDs) and antidepressants.
  • one or more agents are selected from the group consisting of buprenorphine, naloxone, methadone, levomethadyl acerate, L-alpha acetylmethadol (LAAM), hydroxyzine, diphenoxylate, atropine, chlordiazepoxide, carbamazepine, mianserin, benzodiazepine, phenoziazine, disulfiram, acamprosate, topiramate, ondansetron, sertraline, bupropion, amantadine, amiloride, isradipine, tiagabine, baclofen, propranolol, tricyclic antidepressants, desipramine, carbamazepine,
  • the invention provides a composition or combination effective for treating acute pain.
  • the composition comprises a combination of: (i) ibudilast, (ii) an opioid analgesic (e.g ., morphine, oxycodone, or related opioids), and (iii) optionally one or more additional agents effective for treating acute pain, wherein each of the components is either contained in a single composition or dosage form (such as in an admixture), or is present as a discrete or separate entity ( e.g ., in a kit).
  • a composition of the invention may optionally include one or more pharmaceutically acceptable excipients.
  • kits comprising ibudilast, for the treatment of acute pain, and additional agents effective for treating acute pain, for simultaneous, sequential or separate use, as well as instructions for use of the agents.
  • “Pharmaceutically acceptable excipient or carrier” refers to an excipient that may optionally be included in the compositions of the invention and that causes no significant adverse toxicological effects to the patient.
  • “Pharmaceutically acceptable salt” includes, but is not limited to, amino acid salts, salts prepared with inorganic acids, such as chloride, sulfate, phosphate, diphosphate, hydrobromide, and nitrate salts, or salts prepared with an organic acid, such as malate, maleate, fumarate, tartrate, succinate, ethylsuccinate, citrate, acetate, lactate, methanesulfonate, benzoate, ascorbate, para-toluenesulfonate, palmoate, salicylate and stearate, as well as estolate, gluceptate and lactobionate salts.
  • salts containing pharmaceutically acceptable cations include, but are not limited to, sodium, potassium, calcium, aluminum, lithium, and ammonium (including substituted ammonium).
  • Active molecule or “active agent” as described herein includes any agent, drug, compound, composition of matter or mixture which provides some pharmacologic, often beneficial, effect that can be demonstrated in-vivo or in vitro. This includes foods, food supplements, nutrients, nutriceuticals, drugs, vaccines, antibodies, vitamins, and other beneficial agents. As used herein, the terms further include any physiologically or pharmacologically active substance that produces a localized or systemic effect in a patient.
  • acute pain or “subchronic pain” is meant pain resulting from injury to the body and activation of nociceptive transducers at the site of local tissue damage in a subject. Such pain may result from any physical trauma, injury, surgery, or disease (e.g ., post-operative pain, back pain, bums, cancer or other pathological lesions).
  • Acute pain refers to pain that occurs immediately following injury.
  • Subchronic pain refers to prolonged pain following injury that may last a few days, weeks, or months until healing occurs.
  • Nociception is defined herein as pain sense.
  • Nociceptor herein refers to a structure that mediates nociception. The nociception may be the result of a physical stimulus, such as, mechanical, electrical, thermal, or a chemical stimulus. Nociceptors are present in virtually all tissues of the body.
  • Analgesia is defined herein as the relief of pain without the loss of consciousness.
  • An “analgesic” is an agent or drug useful for relieving pain, again, without the loss of consciousness.
  • central nervous system includes all cells and tissue of the brain and spinal cord of a vertebrate. Thus, the term includes, but is not limited to, neuronal cells, glial cells (astrocytes, microglia, oligodendrocytes), cerebrospinal fluid (CSF), interstitial spaces and the like.
  • glial cells astrocytes, microglia, oligodendrocytes
  • CSF cerebrospinal fluid
  • Glial cells refer to various cells of the CNS also known as microglia, astrocytes, and oligodendrocytes.
  • subject refers to a vertebrate, preferably a mammal.
  • Mammals include, but are not limited to, murines, rodents, simians, humans, farm animals, sport animals and pets.
  • an effective amount or “pharmaceutically effective amount” of a composition or agent refers to a nontoxic but sufficient amount of the composition to provide the desired response, such as suppression of glial activation in a subject, and optionally, a corresponding therapeutic effect, such as preventing, diminishing, or eliminating pain in a subject.
  • the exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the condition being treated, the particular drug or drugs employed, mode of administration, and the like. An appropriate "effective” amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation.
  • Treatment or “treating” acute or subchronic pain includes: (1) preventing pain, i.e. causing pain not To develop or to occur with less intensity in a subject that may be exposed to or predisposed to pain but does not yet experience or display pain, (2) inhibiting pain, i.e ., arresting the development or reversing pain, or (3) relieving pain, i.e ., decreasing the amount of pain experienced by the subject.
  • ibudilast By “therapeutically effective dose or amount” of ibudilast is intended an amount that, when ibudilast is administered as described herein, brings about a positive therapeutic response in treatment of acute or subchronic pain, such as preventing, diminishing, or eliminating pain in a subject.
  • the present invention is based on the discovery of a novel therapeutic methodology for safely and effectively treating acute pain using a PDE4 inhibitor, such as ibudilast, in combination with an opioid analgesic.
  • a PDE4 inhibitor such as ibudilast
  • ibudilast potentiates opioid-induced analgesic activity, thus, enhancing opioid-mediated pain relief in a subject.
  • the use of a PDE inhibitor in combination with an opioid analgesic may permit a reduction in the opioid dosage needed to alieviate pain in a subject relative to the amount needed to produce an analgesic effect without administration of the PDE inhibitor, and accordingly, prevent or diminish adverse opioid side-effects, such as opioid dependence and the symptoms of opioid withdrawal that often accompany treatment with opioids.
  • the invention relates to the use of a PDE4- inhibitor, such as ibudilast, in combination with an opioid analgesic to treat acute pain in a subject.
  • Ibudilast has been shown in the present application to potentiate opioid analgesic efficacy.
  • an opioid analgesic e.g ., morphine or oxycodone
  • ibudilast is identified, for the first time, as a potentiator of opioid-induced acute analgesic activity.
  • Exemplary PDE inhibitors other than ibudilast that may also be used to potentiate opioid-induced analgesic activity include, but are not limited to, Rolipram, Arofylline, Roflumilast, Pentoxyfylline, or Propentofylline.
  • Acute pain results from injury to the body and activation of nociceptive transducers at the site of local tissue damage in a subject. Such pain may result from e.g ., physical trauma, surgery, back spasms, dental procedures, such as molar extraction, bums, cancer, or other pathological lesions associated with disease.
  • the methods of the invention are applicable to acute pain occurring immediately following an injury.
  • a therapeutically effective amount of a PDE4 inhibitor and a therapeutically effective amount of an opioid analgesic are administered to a subject in combination therapy with one or more other agents for treating pain.
  • agents include, but are not limited to, analgesics, NSAIDs, benzodiazepines, and antidepressants.
  • agents include, but are not limited to, buprenorphine, naloxone, methadone, levomethadyl acetate, L-alpha acetylmethadol (LAAM), hydroxyzine, diphenoxylate, atropine, chlordiazepoxide, carbamazepine, mianserin, benzodiazepine, phenoziazine, disulfiram, acamprosate, topiramate, ondansetron, sertraline, bupropion, amantadine, amiloride, isradipine, tiagabine, baclofen, propranolol, tricyclic antidepressants, desipramine, carbamazepine, valproate, lamotrigine, doxepin, fluoxetine, imipramine, moclobemide, nortriptyline, paroxetine, sertraline, tryptophan, venlafaxine, trazodone, quetiapin
  • Ibudilast is a small molecule drug (molecular weight of 230.3) having the structure shown below.
  • Ibudilast is also found under ChemBank ID 3227, CAS # 50847-11-5, and Beilstein Handbook Reference No. 5-24-03-00396. Its molecular formula corresponds to [C 14 H 18 N 2 O]. Ibudilast is also known by various chemical names which include 2-methyl-1-(2-(1-methylethyl)pyrazolo(1,5-a)pyridin-3-yl)1-propanone; 3-isobutyryl-2-isopropylpyrazolo(1,5-a)pyridine]; and 1-(2-isopropyl-pyrazolo[1,5-a]pyridin-3-yl)-2-methyl-propan-1-one.
  • Ibudilast examples include Ibudilastum (Latin), BRN 0656579, KC-404, and the brand name Ketas ® .
  • Ibudilast as referred to herein, is meant to include any and all pharmaceutically acceptable salt forms thereof, prodrug forms ( e.g ., the corresponding ketal), and the like, as appropriate for use in its intended formulation for administration.
  • Ibudilast is a non-selective nucleotide phosphodiesterase (PDE) inhibitor (most active against PDE 3, PDE4, PDE 10, and PDE 11 ( Gibson ct al. (2006) Eur. J. Pharmacology 538:39-42 )), and has also been reported to have LTD4 and PAF antagonistic activities. Its profile appears effectively anti-inflammatory and unique in comparison to other PDE inhibitors and anti-inflammatory agents. PDEs catalyze the hydrolysis of the phosphoester bond on the 3'-carbon to yield the corresponding 5'-nucleotide monophosphare. Thus, they regulate the cellular concentrations of cyclic nucleotides.
  • PDE non-selective nucleotide phosphodiesterase
  • PDE1 Ca 2+ /calmodulin-dependent PDEs
  • PDE2 cGMP-stimulated PDEs
  • PDE3 cGMP-inhibited PDEs
  • PDE4 cAMP-specific PDEs
  • PDEs5 cAMP-specific PDEs
  • PDE6 high affinity, cAMP-specific PDEs
  • PDE8 high affinity cGMP-specific PDEs
  • PDE9 mixed cAMP and cGMP PDEs
  • any one or more of the herein-described drugs in particular ibudiiast, is meant to encompass, where applicable, any and all enanriomers, mixtures of enantiomers including racemic mixtures, prodrugs, pharmaceutically acceptable salt forms, hydrates ( e.g ., monohydrates, dihydrates, etc.), different physical forms ( e.g ., crystalline solids, amorphous solids), metabolites, and the like.
  • compositions of the invention may optionally contain one or more additional components as described below.
  • compositions of the invention for treating acute pain may further comprise one or more pharmaceutically acceptable excipients or carriers.
  • excipients include, without limitation, polyethylene glycol (PEG), hydrogenated castor oil (HCO), cremophors, carbohydrates, starches ( e.g ., corn starch), inorganic salts, antimicrobial agents, antioxidants, binders/fillers, surfactants, lubricants ( e.g ., calcium or magnesium stearate), glidants such as talc, disintegrants, diluents, buffers, acids, bases, film coats, combinations thereof, and the like.
  • a composition of the invention may include one or more carbohydrates such as a sugar, a derivatized sugar such as an alditol, aldonic acid, an esterified sugar, and/or a sugar polymer.
  • carbohydrate excipients include, for example: monosaccharides, such as fructose, maltose, galactose, glucose, D-mannose, sorbose, and the like; disaccharides, such as lactose, sucrose, trehalose, cellobiose, and the like; polysaccharides, such as raffinose, melezitose, maltodextrins, dextrans, starches, and the like; and alditols, such as mannitol, xylitol, maltitol, lactitol, xylitol, sorbitol (glucitol), pyranosyl sorbitol, myoinositol, and the like
  • compositions of the invention are potato and corn-based starches such as sodium starch glycolate and directly compressible modified starch.
  • excipients include inorganic salt or buffers such as citric acid, sodium chloride, potassium chloride, sodium sulfate, potassium nitrate, sodium phosphate monobasic, sodium phosphate dibasic, and combinations thereof.
  • a composition comprising a PDE4 inhibitor may also include an antimicrobial agent, e.g ., for preventing or deterring microbial growth.
  • antimicrobial agents suitable for the present invention include benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetylpyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, phenylmercuric nitrate, thimersol, and combinations thereof.
  • a composition comprising a PDE4 inhibitor may also contain one or more antioxidants.
  • Antioxidants are used to prevent oxidation, thereby preventing the deterioration of the drug(s) or other components of the preparation.
  • Suitable antioxidants for use in the present invention include, for example, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, hypophosphorous acid, monothioglycerol, propyl gallate, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite, and combinations thereof.
  • Additional excipients include surfactants such as polysorbates, e.g ., TWEEN 20 and TWEEN 80, and pluronics such as F68 and F88 (both of which are available from
  • sorbitan esters e.g ., phospholipids such as lecithin and other phosphatidylcholines, and phosphatidyluthanolamines
  • lipids e.g ., phospholipids such as lecithin and other phosphatidylcholines, and phosphatidyluthanolamines
  • fatty acids and fatty esters e.g ., steadylcholines, and phosphatidyluthanolamines
  • steroids e.g phospholipids such as lecithin and other phosphatidylcholines, and phosphatidyluthanolamines
  • chelating agents such as EDTA, zinc and other such suitable cations.
  • composition comprising a PDE4 inhibitor may optionally include one or more acids or bases.
  • acids that can be used include those acids selected from the group consisting of hydrochloric acid, acetic acid, phosphoric acid, citric acid, malic acid, lactic acid, formic acid, trichloroacetic acid, nitric acid, perchloric acid, phosphoric acid, sulfuric acid, fumaric acid, and combinations thereof.
  • Suitable bases include, without limitation, bases selected from the group consisting of sodium hydroxide, sodium acetate, ammonium hydroxide, potassium hydroxide, ammonium acetate, potassium acetate, sodium phosphate, potassium phosphate, sodium citrate, sodium formate, sodium sulfate, potassium sulfate, potassium fumarate, and combinations thereof.
  • the amount of any individual excipient in the composition will vary depending on the role of the excipient, the dosage requirements of the active agent components, and particular needs of the composition. Typically, the optimal amount of any individual excipient is determined through routine experimentation, i.e ., by preparing compositions containing varying amounts of the excipient (ranging from low to high), examining the stability and other parameters, and then determining the range at which optimal performance is attained with no significant adverse effects.
  • the excipient will be present in the composition in an amount of about 1% to about 99% by weight, preferably from about 5% to about 98% by weight, more preferably from about 15 to about 95% by weight of the excipient.
  • the amount of excipient present in a 3,4,6-substituted pyridazine composition is selected from the following: at least about 2%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, or even 95% by weight.
  • the formulation (or kit) in accordance with the invention may contain, in addition to a PDE4 inhibitor, one or more opioid analgesics, and optionally additional active agents effective in treating acute or subchronic pain, including, but not limited to, other analgesics, NSAIDs, benzodiazepines and antidepressants.
  • Such actives include morphine, oxycodone, and related opiates, buprenorphine, naloxone, methadone, levomethadyl acetate, L-alpha acetylmethadol (LAAM), hydroxyzine, diphenoxylate, atropine, chlordiazepoxide, carbamazepine, mianserin, benzodiazepine, phenoziazine, disulfiram, acamprosate, topiramate, ondansetron, sertraline, bupropion, amantadine, amiloride, isradipine, tiagabine, baclofen, propranolol, tricyclic antidepressants, desipramine, carbamazepine, valproate, lamotrigine, doxepin, fluoxetine, imipramine, moclobemide, nortriptyline, paroxetine, sertraline, tryptophan, venlafaxine, t
  • opiate receptors that are widely distributed throughout the brain and body. Once an opiate reaches the brain, it quickly activates the opiate receptors found in many brain regions and produces an effect that correlates with the area of the brain involved.
  • opiate receptors There are several types of opiate receptors, including the delta, mu, and kappa receptors. Opiates and endorphins function to block pain signals by binding to the mu receptor site.
  • Gabapentin also known as Neurontin®, is Structurally related to the neurotransmitter GABA. Although structurally related to GABA, gabapenrin does not interact with GABA receptors, is nor converted metabolically into GABA or a GABA agonist, and is not an inhibitor of GABA uptake or degradation. Gabapentin has no activity at GABAA or GABAB receptors of GABA uptake carriers of the brain, but instead interacts with a high-affinity binding site in brain membranes (an auxiliary subunit of voltage-sensitive Ca 2+ channels). The exact mechanism of action is unknown, only that its physiological site of action is the brain. The structure of gabapentin allows it to pass freely through the blood-brain barrier.
  • gabapentin has many pharmacological actions including modulating the action of the GABA synthetic enzyme, increasing non-synaptic GABA responses from neural tissue, and reduction of the release of several mono-amine neurotransmitters.
  • Daily dosages of gabapentin typically range from about 600 to 2400 mg/day, more preferably from about 900 to 1800 mg/day, and are administered in divided doses, for example, three times a day.
  • Conventional unit dosage forms are 300 or 400 mg capsules or 600 or 800 mg tablets.
  • the active agent, memantine is a receptor antagonist. Memantine is believed to function as a low to moderate affinity uncompetitive (open-channel) NMDA receptor antagonist which binds to the NMDA receptor-operated cation channels. Recommended daily dosage amounts typically range from about 5 mg to 20 mg.
  • the cannabinoids e.g. , tetrahydrocannabinol
  • CB 1 receptors are found in brain and peripheral tissues; CB 1 receptors are present in high quantities in the central nervous system, exceeding the levels of almost all neurotransmitter receptors.
  • An additional cannabinoid receptor subtype termed 'CB2' has also been identified. See, e.g. , Martin, B.R., et al., The Journal of Supportive Oncology, Vol. 2, Number 4, July/August 2004 .
  • tramadol Although its mechanism of action has not yet been fully elucidated, the opioid, tramadol, is believed to work through modulation of the GABAergic, noradrenergic and serotonergic systems. Tramadol, and its metabolite, known as M1, have been found to bind to ⁇ -opioid receptors (thus exerting its effect on GABAergic transmission), and to inhibit re-uptake of 5-HT and noradrenaline. The second mechanism is believed to contribute since the analgesic effects of tramadol are not fully antagonised by the ⁇ -opioid receptor antagonist naloxone. Typical daily dosages range from about 50 to 100 milligrams every 4 to 6 hours, with a total daily dosage not to exceed 400 milligrams.
  • Lamotrigine is a phenyltriazine that stabilizes neuronal membranes by blocking voltage-sensitive sodium channels, which inhibit glutamate and aspartate (excitatory amino acid neurotransmitter) release.
  • the daily dosage of lamotrigine typically ranges from 25 milligrams per day to 500 mg per day. Typical daily dosage amounts include 50 mg per day, 100 mg per day, 150 mg per day, 200 mg per day, 300 mg per day, and 500 mgs per day, not exceed 700 mgs per day.
  • Carbamazepine acts by blocking voltage-sensitive sodium channels. Typical adult dosage amounts range from 100-200 milligrams one or two times daily, to an increased dosage of 800-1200 milligrams daily generally administered in 2-3 divided doses.
  • Duloxetine is a potent inhibitor of neuronal uptake of serotonin and norephinephrine and a weak inhibitor of dopamine re-uptake. Typical daily dosage amounts range from about 40 to 60 milligrams once daily, or 20 to 30 milligrams twice daily.
  • Milnacipran acts as a serotonin and norepinephrine reuptake inhibitor.
  • Daily dosage amounts typically range from about 50 to 100 milligrams once or twice daily.
  • dosage amounts provided above are meant to be merely guidelines; the precise amount of a secondary active agent to be administered during combination therapy with ibudilast will, of course, be adjusted accordingly and will depend upon factors such as inrendcd patient population, the particular acute or subchronic pain symptom or condition to be treated, potential synergies between the active agents administered, and the like, and will readily be determined by one skilled in the art based upon the guidance provided herein.
  • compositions may also be formulated in order to improve stability and extend the half-life of the PDE4 inhibitor.
  • the PDE inhibitor may be delivered in a sustained-release formulation.
  • Controlled or sustained-release formulations are prepared by incorporating ibudilast into a carrier or vehicle such as liposomes, nonresorbable impermeable polymers such as ethylenevinyl acetate copolymers and Hytrel® copolymers. swellable polymers such as hydrogels, or resorbable polymers such as collagen and certain polyacids or polyesters such as those used to make resorbable sutures.
  • the PDE, inhibitor can be encapsulated, adsorbed to, or associated with, particulate carriers.
  • particulate carriers include those derived from polymethyl methacrylate polymers, as well as microparticles derived from poly(lactides) and poly(lactide-co-glycolides), known as PLG. See, e.g., Jeffery et al., Pharm. Res. (1993) 10:62-368 ; and McGee et al., J. Microencap. (1996 ).
  • compositions comprising a PDE inhibitor described herein encompass all types of formulations, and in particular, those that are suited for systemic or intrathecal administration.
  • Oral dosage forms include tablets, lozenges, capsules, syrups, oral suspensions, emulsions, granules, and pellets.
  • Alternative formulations include aerosols, transdermal patches, gels, creams, ointments, suppositories, powders or lyophilates that can be reconstituted, as well as liquids.
  • Suitable diluents for reconstituting solid compositions include bacteriostatic water for injection, dextrose 5% in water, phosphate-buffered saline, Ringer's solution, saline, sterile water, deionized water, and combinations thereof.
  • bacteriostatic water for injection dextrose 5% in water
  • phosphate-buffered saline Ringer's solution
  • saline sterile water
  • deionized water deionized water
  • a composition comprising a PDE4 inhibitor is one suited for oral administration.
  • tablets can be made by compression or molding, optionally with one or more accessory ingredients or additives.
  • Compressed tablets are prepared, for example, by compressing in a suitable tabletting machine, the active ingredients in a free-flowing form such as a powder or granules, optionally mixed with a binder (e.g. , povidone, gelatin, hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (e.g ., sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymethyl cellulose) and/or surface-active or dispersing agent.
  • a binder e.g., povidone, gelatin, hydroxypropylmethyl cellulose
  • lubricant e.g., povidone, gelatin, hydroxypropylmethyl cellulose
  • inert diluent e.g., preservative
  • disintegrant e.g ., sodium starch glycolate, cross-linked
  • Molded tablets are made, for example, by molding in a suitable tabletting machine, a mixture of powdered compounds moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored, and may be formulated so as to provide slow or controlled release of the active ingredients, using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile.
  • Tablets may optionally be provided with a coating, such as a thin film, sugar coating, or an enteric coating to provide release in parts of the gut other than the stomach. Processes, equipment, and toll manufacturers for tablet and capsule making are well-known in the art.
  • Formulations for topical administration in the mouth include lozenges comprising the active ingredients, generally in a flavored base such as sucrose and acacia or tragacanth and pastilles comprising the active ingredients in an inert base such as gelatin and glycerin or sucrose and acacia.
  • a pharmaceutical composition for topical administration may also be formulated as an ointment, cream, suspension, lotion, powder, solution, paste, gel, spray, aerosol or oil.
  • the formulation may be in the form of a patch ( e.g. , a transdermal patch) or a dressing such as a bandage or adhesive plaster impregnated with active ingredients and optionally one or more excipients or diluents.
  • Topical formulations may additionally include a compound that enhances absorption or penetration of the ingredients through the skin or other affected areas, such as dimethylsulfoxidem bisabolol, oleic acid, isopropyl myristate, and D-limonene, to name a few.
  • the oily phase is constituted from known ingredients in a known manner. While this phase may comprise merely an emulsifier (otherwise known as an emulgent), it desirably comprises a mixture of at least one emulsifier with a fat and/or an oil. Preferably, a hydrophilic emulsifier is included together with a lipophilic emulsifier that acts as a stabilizer. Together, the emulsifier(s) with or without stabilizer(s) make up the so-called emulsifying wax, and the wax together with the oil and/or fat make up the so-called emulsifying ointment base which forms the oily dispersed phase of cream formulations.
  • Illustrative emulgents and emulsion stabilizers include TWEEN 60, SPAN 80, cetostearyl alcohol, myristyl alcohol, glyceryl monostearate and sodium lauryl sulfate.
  • Formulations for rectal administration are typically in the form of a suppository with a suitable base comprising, for example, cocoa butter or a salicylate.
  • Formulations suitable for vaginal administration generally take the form of a suppository, tampon, cream, gel, paste, foam or spray.
  • Formulations suitable for nasal administration include a coarse powder having a particle size, for example, in the range of about 20 to about 500 microns. Such a formulation is typically administered by rapid inhalation through the nasal passage, e.g. , from a container of the powder held in proximity to the nose.
  • a formulation for nasal delivery may be in the form of a liquid, e.g ., a nasal spray or nasal drops.
  • Aerosolizable formulations for inhalation may be in dry powder form (e.g ., suitable for administration by a dry powder inhaler), or, alternatively, may be in liquid form, e.g ., for use in a nebulizer.
  • Nebulizers for delivering an aerosolized solution include the AERxTM (Aradigm), the Ultravent® (Mallinkrodt), and the Acorn II® (Marquest Medical Products).
  • a composition of the invention may also be delivered using a pressurized, metered dose inhaler (MDI), e.g ., the Ventolin® metered dose inhaler, containing a solution or suspension of a combination of drugs as described herein in a pharmaceutically inert liquid propellant, e.g ., a chlorofluorocarbon or fluorocarbon.
  • MDI pressurized, metered dose inhaler
  • a pharmaceutically inert liquid propellant e.g ., a chlorofluorocarbon or fluorocarbon.
  • Formulations suitable for parenteral administration include aqueous and nonaqueous isotonic sterile solutions suitable for injection, as well as aqueous and nonaqueous sterile suspensions.
  • Parenteral formulations are optionally contained in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, water for injections, immediately prior to use.
  • sterile liquid carrier for example, water for injections
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the types previously described.
  • a formulation for use with the invention may also be a sustained release formulation, such that each of the drug components is released or absorbed slowly over time, when compared to a non-sustained release formulation.
  • Sustained release formulations may employ pro-drug forms of the active agent, delayed-release drug delivery systems such as liposomes or polymer matrices, hydrogels, or covalent attachment of a polymer such as polyethylene glycol to the active agent.
  • the formulations may optionally include other agents conventional in the pharmaceutical arts and particular type of formulation being employed, for example, for oral administration forms, the composition for oral administration may also include additional agents as sweeteners, thickeners or flavoring agents.
  • compositions comprising a PDE4 inhibitor may also be prepared in a form suitable for veterinary applications.
  • kits containing a composition comprising a PDE4 inhibitor (e.g ., ibudilast), accompanied by instructions for use, e.g ., in treating acute pain.
  • a PDE4 inhibitor e.g ., ibudilast
  • the kit may optionally contain an opioid analgesic (e.g ., morphine or oxycodone) or one or more other agents for treating pain.
  • the kit comprises a PDE4 inhibitor in addition to each of the drugs making up the composition of the invention, along with instructions for use.
  • a PDE inhibitor and one or more opioids or other agents may be present in the same or separate compositions.
  • the drug components may be packaged in any manner suitable for administration, so long as the packaging, when considered along with the instructions for administration, clearly indicates the manner in which each of the drug components is to be administered.
  • kits comprising ibudilast and morphine
  • the kit may be organized by any appropriate time period, such as by day.
  • a representative kit may comprise unit dosages of each of ibudilast and morphine. If each of the drugs is to be administered twice daily, then the kit may contain, corresponding to Day 1, two rows of unit dosage forms of each of ibudilast and morphine, along with instructions for the timing of administration.
  • the kit may contain, corresponding to Day 1, two rows of unit dosage forms of each of ibudilast and morphine, along with instructions for the timing of administration.
  • one or more of the drugs differs in the timing or quantity of unit dosage form to be administered in comparison to the other drug members of the combination, then such would be reflected in the packaging and instructions.
  • the packaging may be in any form commonly employed for the packaging of pharmaceuticals, and may utilize any of a number of features such as different colors, wrapping, ramper-resistant packaging, blister paks, dessicants, and the like.
  • the present invention encompasses a method of treating a mammalian subject suffering from acute pain by administering therapeutically effective dosages of a PDE4 inhibitor and an opioid analgesic. Such administering is effective to decrease the amount of pain experienced by the subject, i.e. , to result in significant attenuation or elimination of acute pain.
  • Therapeutic amounts can be empirically determined and will vary with the particular condition being treated, the subject, the specific opioid analgesic, and the particular efficacy and toxicity of each of the active agents contained in the composition.
  • the actual dose to be administered will vary depending upon the age, weight, and general condition of the subject as well as the severity of the condition being treated, the judgment of the health care professional, and particular mode of administration.
  • the method of the invention may, in certain instances, comprise a step of selecting a subject experiencing acute or subchronic pain prior to administering thereto ibudilast and an analgesic opioid.
  • Such subjects are typically selected from those suffering from physical trauma associated with injury, surgery, or disease, (e.g., post-operative pain, back spasms, bums, cancer, or pathological lesions).
  • the method of the invemion may be effective to not only significantly attenuate acute pain, but to even reverse it, such that the resulting pain relief is long-lasting.
  • the administering of a PDE4 inhibitor in combination with an opioid analgesic as described herein may be effective to result in sustained attenuation of acute pain for an overnight duration.
  • therapeutically effective doses of a PDE inhibitor and an opioid analgesic may be effective to treat acute pain for a duration of up to at least 4 hours, 6 hours, 8 hours, 10 hours, 12 hours, 16 hours, 18 hours or even 20 hours or greater.
  • the PDE inhibitor and an opioid analgesic may also be administered in combination with one or more additional agents effective for treating acute pain.
  • agents include other analgesics, non-steroidal anti-inflammatory drugs (NSAIDs), benzodiazepines and antidepressants.
  • Preferred methods of delivery of therapeutic formulations comprising a PDE4 inhibitor for the treatment of acute pain include systemic and localized delivery, i.e ., directly into the central nervous system.
  • routes of administration include but are not limited to, oral, intra-arterial, intrathecal, intraspinal, intramuscular, subcutaneous, intraperitoneal, intravenous, intranasal, and inhalation routes.
  • a formulation containing a PDE4 inhibitor of the present invention may be administered for therapy by any suitable route, including without limitation, oral, rectal, nasal, topical (including transdermal, aerosol, buccal and sublingual), vaginal, parenteral (including subcutaneous, intramuscular, intravenous and intradermal), intrathecal, and pulmonary.
  • suitable route including without limitation, oral, rectal, nasal, topical (including transdermal, aerosol, buccal and sublingual), vaginal, parenteral (including subcutaneous, intramuscular, intravenous and intradermal), intrathecal, and pulmonary.
  • the preferred route will, of course, vary with the condition and age of the recipient, the particular neuralgia-associated syndrome being treated, and the specific combination of drugs employed.
  • One preferred mode of administration for delivery of a PDE4 inhibitor is directly to neural tissue such as peripheral nerves, the retina, dorsal root ganglia, neuromuscular junction, as well as the CNS, e.g. , to target spinal cord glial cells by injection into, e.g ., the ventricular region, as well as to the striatum (e.g ., the caudate nucleus or putamen of the striatum), spinal cord and neuromuscular junction, with a needle, catheter or related device, using neurosurgical techniques known in the art, such as by stereotactic injection (see, e.g., Stein et al., J. Virol.
  • a particularly preferred method for targeting spinal cord glia is by intrathecal delivery, rather than into the cord tissue itself.
  • compositions comprising a PDE4 inhibitor of the invention is by delivery to dorsal root ganglia (DRG) neurons, e.g ., by injection into the epidural space with subsequent diffusion to DRG.
  • DRG dorsal root ganglia
  • an ibudilast-based composition can be delivered via intrathecal cannulation under conditions where ibudilast is diffused to DRG. See, e.g., Chiang et al., Acta Anaesthesiol. Sin. (2000) 38:31-36 ; Jain, K.K., Expert Opin. Investig. Drugs (2000) 9:2403-2410 .
  • CED convection-enhanced delivery
  • the PDE inhibitor can be delivered to many cells over large areas of the CNS.
  • Any convection-enhanced delivery device may be appropriate for delivery of ibudilast.
  • the device is an osmotic pump or an infusion pump. Both osmotic and infusion pumps are commercially available from a variety of suppliers, for example Alzet Corporation, Hamilton Corporation, Alza, Inc., Palo Also, California).
  • a composition comprising a PDE inhibitor or glial attenuator of the invention is delivered via CED devices as follows.
  • a catheter, cannula or other injection device is inserted into CNS tissue in the chosen subject.
  • Stereotactic maps and positioning devices are available, for example from ASI Instruments, Warren, MI. Positioning may also be conducted by using anatomical maps obtained by CT and/or MRI imaging to help guide the injection device to the chosen target.
  • anatomical maps obtained by CT and/or MRI imaging to help guide the injection device to the chosen target.
  • a composition comprising a PDE4 inhibitor when comprising more than one active agent, may be administered as a single combination composition comprising a combination of the PDE4 inhibitor and at least one additional active agent effective in the treatment of acute or subchronic pain.
  • the combination of the invention is administered as separate dosage forms.
  • the PDE4 inhibitor and each of the additional active agents may be administered simultaneously, sequentially in any order, or separately.
  • Therapeutic amounts can be empirically determined by those skilled in the art and will vary with the particular condition being treated, the subject, the particular opioid employed, and the efficacy and toxicity of each of the active agents contained in the composition.
  • the actual dose to be administered will vary depending upon the age, weight, and general condition of the subject as well as the severity of the condition being treated, the judgment of the health care professional, and the particular combination of opioid, PDE inhibitor, and any other agents being administered.
  • Therapeutically effective amounts can be determined by those skilled in the art, and will be adjusted to the requirements of each particular case.
  • a therapeutically effective amount of a PDE inhibitor will range from a total daily dosage, for example in humans, of about 0.1 and 500 mg/day, more preferably, in an amount between I and 200 mg/day, I and 100 mg/day, I and 40 mg/day, or 1 and 20 mg/day, administered as either a single dosage or as multiple dosages.
  • administration can be one, two, or three times daily, or even more, for a time course of one day to several days, weeks, months, and even years, and may even be for the life of the patient.
  • Intermittent dosing may also be employed, e.g. , in response to acute or subchronic pain, with a maximal dose not to be exceeded as recommended by the practicing physician.
  • Illustrative dosing regimes will last a period of at least about a week, from about 1-4 weeks, from 1-3 months, from 1-6 months, from 1-50 weeks, from 1-12 months, or longer,
  • a PDE inhibitor in combination with an opioid analgesic to treat acute pain can be evaluated by any of the standard pain models known in the art. Examples of such models are as follows.
  • Tail Flick Model The tail-flick test ( D'Amour et al. (1941) J. Pharmacol. Exp. and Ther. 72:74-79 ) is a model of acute pain.
  • a towel-wrapped rat is placed on a test stage such that a focused light source beams on the dorsal surface of the rat's tail.
  • a photosensor is present on the test stage located opposite the light source and below the rat's tail. To begin the test, the rat's tail blocks the light, thus preventing the light reaching the photosensor. Latency measurement begins with the activation of the light source. When a rat moves or flicks its tail, the photosensor detects the light source and stops the measurement.
  • the test measures the period of time (duration) that the rat's tail remains immobile (latent). Rats are tested prior to administration thereto of a compound of interest and then at various times after such administration.
  • the light source is set to an intensity that produced a tail response latency of about 3 seconds when applied to the tails of rats to which no compound has been administered.
  • Rat Tail Immersion Model The rat tail immersion assay is also a model of acute pain. A rat is loosely held in hand while covered with a small folded thin cotton towel with its tail exposed. The tip of the tail is dipped into a, e.g., 52°C water bath to a depth of two inches. The rat responds by either wiggling of the tail or withdrawal of the tail from the water; either response is scored as the behavioral end-point. Rats are tested for a tail response latency (TRL) score prior to administration thereto of a compound of interest and then retested for TRL at various times after such administration.
  • TRL tail response latency
  • Carrageenan-induced Paw Hyperalgesia Model The carrageenan paw hyperalgesia test is a model of inflammatory pain. A subcutaneous injection of carrageenan is made into the left hindpaws of rats. The rats are treated with a selected agent before, e.g., 30 minutes, the carrageenan injection or after, e.g., two hours after, the carrageenan injection. Paw pressure sensitivity for each animal is tested with an analgesymeter three hours after the carrageenan injection. See, Randall et al. (1957) Arch. Int. Pharmacodyn. 111:409-419 .
  • Formalin Behavioral Response Model The formalin test is a model of acute, persistent pain. Response to formalin treatment is biphasic ( Dubuisson et al. (1977) Pain 4:161-174 ). The Phase I response is indicative of a pure nociceptive response to the irritant. Phase 2, typically beginning 20 to 60 minutes following injection of formalin, is thought to reflect increased sensitization of the spinal cord.
  • Von Frey Filament Test The effect of compounds on mechanical allodynia can be determined by the von Frey filament test in rats with a tight ligation of the L-5 spinal nerve: a model of painful peripheral neuropathy. The surgical procedure is performed as described by Kim et al. (1992) Pain 50:355-363 . A calibrated series of von Frey filaments are used to assess mechanical allodynia ( Chaplan et al. (1994) J. Neurosci. Methods 53:55-63 ). Filaments of increasing stiffness are applied perpendicular to the midplantar surface in the sciatic nerve distribution of the left hindpaw. The filaments are slowly depressed until bending occurred and are then held for 4-6 seconds. The filament application order and number of trials were determined by the up-down method of Dixon (Chaplan et al., supra). Flinching and licking of the paw and paw withdrawal on the ligated side are considered positive responses.
  • Chronic Constriction Injury Heat and cold allodynia responses can be evaluated as described below in rats having a chronic constriction injury (CCI).
  • CCI chronic constriction injury
  • a unilateral mononeuropathy is produced in rats using the chronic constriction injury model described in Bennett et al. (1988) Pain 33:87-107 .
  • CCI is produced in anesthetized rats as follows.
  • the lateral aspect of each rat's hind limb is shaved and scrubbed with Nolvasan.
  • an incision is made on the lateral aspect of the hind limb at the mid-thigh level.
  • the biceps femoris is bluntly dissected to expose the sciatic nerve.
  • four loosely tied ligatures for example, Chromic gut 4.0; Ethicon, Johnson and Johnson, Somerville, NJ
  • an identical dissection is performed except that the sciatic nerve is not ligated (sham).
  • the muscle is closed with a continuous suture pattern with, e.g., 4-0 Vicryl (Johnson and Johnson, Somerville, NJ) and the overlying skin is closed with wound clips.
  • the rats are ear-tagged for identification purposes and returned to animal housing.
  • CCI rats are tested for thermal hyperalgesia at least 10 days post-op.
  • the test apparatus consists of an elevated heated (80-82°F) glass platform. Eight rats at a time, representing all testing groups, are confined individually in inverted plastic cages on the glass floor of the platform at least 15 minutes before testing. A radiant heat source placed underneath the glass is aimed at the plantar hind paw of each rat. The application of heat is continued until the paw is withdrawn (withdrawal latency) or the time elapsed is 20 seconds. This trial is also applied to the sham operated leg. Two to four trials are conducted on each paw, alternately, with at least 5 minutes interval between trials. The average of these values represents the withdrawal latency.
  • Cold Allodynia Model The test apparatus and methods of behavioral testing is described in Gogas et al. (1997) Analgesia 3:111-118 .
  • the apparatus for testing cold allodynia in neuropathic (CCI) rats consists of a Plexiglass chamber with a metal plate 6 cm from the bottom of the chamber. The chamber is filled with ice and water to a depth of 2.5 cm above the metal plate, with the temperature of the bath maintained at 0-4°C throughout the test.
  • the Hargreaves Test ( Hargreaves et al., Pain (1998) 32:77-88 ) is also a radiant heat model for pain.
  • CCI rats are tested for thermal hyperalgesia at least 10 days post-op.
  • the test apparatus consists of an elevated heated (80-82°F) glass platform. Eight rats at a time, representing all testing groups, are confined individually in inverted plastic cages on the glass floor of the platform at least 15 minutes before testing.
  • a radiant heat source placed underneath the glass is aimed at the plantar hind paw of each rat. The application of heat is continued until the paw is withdrawn (withdrawal latency) or the time elapsed is 20 seconds. This trial is also applied to the sham operated leg. Two to four trials are conducted on each paw, alternately, with at least 5 minutes interval between trials. The average of these values represents the withdrawal latency.
  • Morphine sulfate and oxycodone hydrochloride were purchased from Sigma (St. Louis, MO, USA). Morphine and oxycodone were administered subcutaneously at 4 mg/kg in a dose volume of 1 ml/kg in sterile injection normal saline. Ibudilast was supplied by Avigen (Alameda, CA, USA). Ibudilast was administered intraperitoneally at 7.5 mg/kg in a dose volume of 2.5 ml/kg in 35% PEG: 65% injection saline. Morphine and oxycodone doses reported are as free base concentrations.
  • Hargreaves tests for analgesia and hyperalgesia Rats received at least three 60 min habituations to the test environment prior to behavioral testing. Thresholds for behavioral response to heat stimuli applied to the plantar surface of each hindpaw and tail were assessed using a modified Hargreaves test (Hargreaves et al., 1988). All testing was conducted blind with respect to group assignment. Briefly, baseline withdrawal values were calculated from an average of 2 consecutive withdrawal latencies of the tail and the left and the right hindpaws, measured at 15-min intervals. Latencies for the high intensity stimulus at baseline ranged from 2 to 3 s, and a cut-off time of 10 s was imposed to avoid tissue damage. Latencies for the low intensity stimulus at baseline ranged from 6 to 7 s, and a cut-off time of 20 s was imposed to avoid tissue damage. The order of paw and tail testing varied randomly.
  • mice Following baseline withdrawal latency assessments (-45 min), animals received ibudilast or vehicle and were tested again 10 min later to determine if ibudilast or vehicle had any effect on withdrawal latencies. At this time (time 0), the animals received a subcutaneous saline injection (1 ml/kg) to control for conditions in Experiment 2 (see below). Animals were then tested every 10 minutes for 80 min alternating high and low intensity Hargreaves tests.
  • animals Following baseline withdrawal latency assessments (-45 min), animals received ibudilast or vehicle (-30 min) and were tested again at -10 min. At time 0, animals received a subcutaneous morphine injection (4 mg/kg in a dose volume of 1 ml/kg in injection saline) and were tested every 10 minutes for 230 min alternating high and low intensity Hargreaves tests.
  • animals Following baseline withdrawal latency assessments (-45 min), animals received ibudilast or vehicle (-30 min) and were tested again at -10 min. At time 0, animals received a subcutaneous oxycodone injection (4 mg/kg in a dose volume of 1 ml/kg in injection saline) and were tested every 10 minutes for 230 min alternating high and low intensity Hargreaves tests.
  • the light source was moved for the 2nd and 3rd trials one cm caudal and rostral of the original trial to avoid tissue damage.
  • Animals were dosed orally with ibudilast (50 mg/kg in vehicle (10% HCO60 + 10% PEG in sterile water) and/or 1.2 mg/kg morphine s.c. formulated in saline for injection. Dose volumes were 1 ml/kg for s.c. administrations (i.e. opioid) and 10 ml/kg for p.o. administrations. Animals were dosed and returned to their cages until 5 minutes before each hourly time point, when they were returned to the glass surface and stimulated with the heat source (3 trials/time point). The ibudilast vehicle was also shown not to have analgesic activity and performed like the saline control. Studies were performed at Avigen Inc. under IACUC regulations.
  • morphine 1.2 mg/kg s.c.
  • ibudilast 50 mg/kg po
  • co-administration of oral ibudilast and s.c. morphine results in a substantial, greater-than-additive increase in tail-flick latency times in the combination treatment group versus either individual therapeutic alone.
  • ibudilast is shown to clearly potentiate morphine-induced analgesia in a classic rat model of analgesia.
  • Ibudilast is used to potentiate the antinociceptive or analgesic efficacy of opioids (e.g. , morphine, oxycodone).
  • opioids e.g. , morphine, oxycodone

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Emergency Medicine (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Pain & Pain Management (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
EP08724934A 2007-01-30 2008-01-29 Methods for treating acute pain Not-in-force EP2131841B1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PL08724934T PL2131841T3 (pl) 2007-01-30 2008-01-29 Sposoby leczenia bólu ostrego

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US89836207P 2007-01-30 2007-01-30
PCT/US2008/001187 WO2008094571A1 (en) 2007-01-30 2008-01-29 Methods for treating acute and subchronic pain

Publications (2)

Publication Number Publication Date
EP2131841A1 EP2131841A1 (en) 2009-12-16
EP2131841B1 true EP2131841B1 (en) 2012-08-01

Family

ID=39345258

Family Applications (1)

Application Number Title Priority Date Filing Date
EP08724934A Not-in-force EP2131841B1 (en) 2007-01-30 2008-01-29 Methods for treating acute pain

Country Status (8)

Country Link
US (1) US20080181876A1 (pl)
EP (1) EP2131841B1 (pl)
DK (1) DK2131841T3 (pl)
ES (1) ES2392116T3 (pl)
HK (1) HK1139856A1 (pl)
PL (1) PL2131841T3 (pl)
PT (1) PT2131841E (pl)
WO (1) WO2008094571A1 (pl)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL1928438T3 (pl) * 2005-09-26 2013-08-30 Avigen Inc Zastosowanie ibudilastu do leczenia uzależnień od środków odurzających
CA2653345A1 (en) 2006-05-31 2007-12-13 Avigen, Inc. Ibudilast for inhibiting macrophage migration inhibitory factor (mif) activity
CA2669463A1 (en) * 2006-11-09 2008-05-15 Avigen, Inc. Method for treating delirium
WO2008137012A1 (en) * 2007-05-03 2008-11-13 Avigen, Inc. Use of a glial attenuator to prevent amplified pain responses caused by glial priming
CA2785056A1 (en) 2009-12-22 2011-07-21 Pondera Biotechnologies, LLC Methods and compositions for treating distress dysfunction and enhancing safety and efficacy of specific medications
US9693949B1 (en) 2015-12-22 2017-07-04 Revogenex Ireland Ltd Intravenous administration of tramadol
US9980900B2 (en) 2015-12-22 2018-05-29 Revogenex Ireland Ltd Intravenous administration of tramadol
EP3743145A4 (en) 2018-01-25 2021-12-15 Cognifisense, Inc. COMBINATORY THERAPEUTIC SYSTEMS AND PROCEDURES
US11977085B1 (en) 2023-09-05 2024-05-07 Elan Ehrlich Date rape drug detection device and method of using same

Family Cites Families (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5229318B2 (pl) 1972-03-30 1977-08-01
US4097483A (en) 1974-11-01 1978-06-27 Kyorin Pharmaceutical Co., Ltd. Pyrazolo 1,5-a!pyridines
JPS6263520A (ja) * 1985-09-14 1987-03-20 Kyorin Pharmaceut Co Ltd 抗リウマチ剤
WO1996041626A1 (en) * 1995-06-12 1996-12-27 G.D. Searle & Co. Compositions comprising a cyclooxygenase-2 inhibitor and a 5-lipoxygenase inhibitor
US20040014761A1 (en) * 1997-10-28 2004-01-22 Place Virgil A. Treatment of female sexual dysfunction with phosphodiesterase inhibitors
ES2326893T3 (es) * 1998-05-27 2009-10-21 Genzyme Corporation Vectores aav para la fabricacion de medicamentos para administracion potenciada por conveccion.
CA2319495A1 (en) * 1998-06-08 1999-12-16 Advanced Medicine, Inc. Multibinding inhibitors of microsomal triglyceride transferase protein
WO2000009127A1 (fr) * 1998-08-10 2000-02-24 Kyorin Pharmaceutical Co., Ltd. Traitements contre la sclerose en plaques
US20020068740A1 (en) * 1999-12-07 2002-06-06 Mylari Banavara L. Combination of aldose reductase inhibitors and antihypertensive agents for the treatment of diabetic complications
US7135495B2 (en) * 2000-03-09 2006-11-14 Ono Pharmaceutical Co., Ltd. Indole derivatives
WO2001074811A2 (en) * 2000-03-30 2001-10-11 Takeda Chemical Industries, Ltd. Substituted 1,3-thiazole compounds, their production and use
GB0008269D0 (en) * 2000-04-05 2000-05-24 Astrazeneca Ab Combination chemotherapy
ATE509904T1 (de) * 2000-09-14 2011-06-15 Mitsubishi Tanabe Pharma Corp Neue amidverbindungen und deren medizinische verwendung
US20020137755A1 (en) * 2000-12-04 2002-09-26 Bilodeau Mark T. Tyrosine kinase inhibitors
DE20020546U1 (de) * 2000-12-04 2002-04-11 Zumtobel Staff Gmbh Fassung für Kompaktleuchtstofflampen
CA2431206C (en) * 2000-12-08 2009-09-01 Takeda Chemical Industries, Ltd. Combination drugs containing anti-sepsis cycloalkene compound
US20040097555A1 (en) * 2000-12-26 2004-05-20 Shinegori Ohkawa Concomitant drugs
EP1402900A1 (en) * 2001-06-11 2004-03-31 Takeda Chemical Industries, Ltd. Medicinal compositions
JP2004538305A (ja) * 2001-07-31 2004-12-24 ファルマシア・アンド・アップジョン・カンパニー 3−ヘテロシクリル−及び3−シクロアルキル−3−アリールオキシプロパンアミンによる慢性疼痛の治療
JP5137289B2 (ja) * 2001-08-03 2013-02-06 武田薬品工業株式会社 安定な乳化組成物
CA2456754A1 (en) * 2001-08-08 2003-02-20 Yuji Iizawa Benzazepine derivative, process for producing the same, and use
CA2457482A1 (en) * 2001-08-09 2003-02-27 Kissei Pharmaceutical Co., Ltd. 5-amidino-n-(2-aminophenethyl)-2-hydroxybenzenesulfonamide derivative, medicinal composition containing the same, and intermediate therefor
US7338939B2 (en) * 2003-09-30 2008-03-04 New River Pharmaceuticals Inc. Abuse-resistant hydrocodone compounds
WO2003022813A1 (fr) * 2001-09-07 2003-03-20 Ono Pharmaceutical Co., Ltd. Derives indole, methode de fabrication et medicaments renfermant lesdits derives en tant que principe actif
CA2464763A1 (en) * 2001-11-09 2003-05-15 Kissei Pharmaceutical Co., Ltd. 5-amidino-2-hydroxybenzenesulfonamide derivatives, medicinal compositions containing the same, medicinal use thereof and intermediates in the production thereof
KR100704215B1 (ko) * 2002-03-05 2007-04-10 오노 야꾸힝 고교 가부시키가이샤 8-아자프로스타글란딘 유도체 화합물 및 그 화합물을 유효성분으로서 함유하는 약제
US20040053842A1 (en) * 2002-07-02 2004-03-18 Pfizer Inc. Methods of treatment with CETP inhibitors and antihypertensive agents
US20050038063A1 (en) * 2002-11-29 2005-02-17 Kenneth Newman Method of treating acute pain with unitary dosage form comprising ibuprofen and oxycodone
CL2004000545A1 (es) * 2003-03-18 2005-01-28 Pharmacia Corp Sa Organizada B Uso de un antagonista de los receptores de aldosterona y un antagonista de receptores de endotelina para el tratamiento o profilaxis de una condicion patologica relacionada con hipertension, disfuncion renal, insulinopatia y enfermedades cardiovascul
CL2004000544A1 (es) * 2003-03-18 2005-01-28 Pharmacia Corp Sa Organizada B Uso de una combinacion farmaceutica, de un antagonista del receptor de aldosterona y un inhibidor de endopeptidasa neutral, util para el tratamiento y prevencion de una condicion patologica relacionada con hipertension, disfuncion renal, insulinopati
WO2004100871A2 (en) * 2003-05-09 2004-11-25 Pharmacia Corporation Combination of an aldosterone receptor antagonist and a renin inhibitor
US7261882B2 (en) * 2003-06-23 2007-08-28 Reagents Of The University Of Colorado Methods for treating neuropathic pain by administering IL-10 polypeptides
TW200517114A (en) * 2003-10-15 2005-06-01 Combinatorx Inc Methods and reagents for the treatment of immunoinflammatory disorders
US7534806B2 (en) * 2004-12-06 2009-05-19 Avigen, Inc. Method for treating neuropathic pain and associated syndromes
MX2007015789A (es) * 2005-06-16 2008-02-15 Mallinckrodt Inc Nueva ruta de sintesis para opiatos de 14-hidroxilo a traves de 1-halo-tebaina o analogos.
PL1928438T3 (pl) * 2005-09-26 2013-08-30 Avigen Inc Zastosowanie ibudilastu do leczenia uzależnień od środków odurzających
CA2653345A1 (en) * 2006-05-31 2007-12-13 Avigen, Inc. Ibudilast for inhibiting macrophage migration inhibitory factor (mif) activity
AU2007258567B2 (en) * 2006-06-06 2012-04-19 Medicinova, Inc. Substituted pyrazolo (1,5-alpha) pyridine compounds and their methods of use
CA2669463A1 (en) * 2006-11-09 2008-05-15 Avigen, Inc. Method for treating delirium

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CAMPOS ADRIANA ROLIM ET AL: "Ketamine-induced potentiation of morphine analgesia in rat tail-flick test: Role of opioid-, alpha(2)-adrenoceptors and ATP-sensitive potassium channels", BIOLOGICAL & PHARMACEUTICAL BULLETIN, vol. 29, no. 1, January 2006 (2006-01-01), pages 86 - 89, ISSN: 0918-6158 *

Also Published As

Publication number Publication date
WO2008094571A1 (en) 2008-08-07
PL2131841T3 (pl) 2013-03-29
PT2131841E (pt) 2012-09-24
ES2392116T3 (es) 2012-12-04
HK1139856A1 (en) 2010-09-30
US20080181876A1 (en) 2008-07-31
DK2131841T3 (da) 2012-10-15
EP2131841A1 (en) 2009-12-16

Similar Documents

Publication Publication Date Title
EP2131841B1 (en) Methods for treating acute pain
JP5411504B2 (ja) 薬物嗜癖および行動嗜癖を治療するためのイブジラストの使用
JP4954085B2 (ja) 神経障害性疼痛及びその関連する症状を治療するためのイブジラスト
EP2187882B1 (en) Treatment of progressive neurodegenerative disease with ibudilast
KR20030040201A (ko) 급성 통증, 만성 통증 및/또는 신경병성 통증 및 편두통을치료하기 위한 약학 조성물
US20080287402A1 (en) Use of a glial attenuator to prevent amplified pain responses caused by glial priming
US20090028816A1 (en) Treatment of depression, psychosis, and anxiety
WO2019157423A1 (en) Methods and dosing regimens using ibudilast and a second agent for cancer therapy
US20070281924A1 (en) MIF inhibitors for treating neuropathic pain and associated syndromes
US20080114027A1 (en) Method for treating delirium
US20230090534A1 (en) Methods of treating glioblastoma multiforme using combination therapy
RU2433825C2 (ru) Способ лечения невропатической боли и связанных с ней синдромов
US20110275664A1 (en) Method for treating drug and behavioral addictions
US20070191365A1 (en) 3,4,6-Substituted pyridazines for treating neuropathic pain and associated syndromes

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20090831

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

RIN1 Information on inventor provided before grant (corrected)

Inventor name: WATKINS, LINDA

Inventor name: JOHNSON, KIRK, W.

Inventor name: HUTCHINSON, MARK

DAX Request for extension of the european patent (deleted)
17Q First examination report despatched

Effective date: 20100702

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1139856

Country of ref document: HK

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

RTI1 Title (correction)

Free format text: METHODS FOR TREATING ACUTE PAIN

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 568267

Country of ref document: AT

Kind code of ref document: T

Effective date: 20120815

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

BECA Be: change of holder's address

Owner name: 1800 GRANT STREET 8TH FLOOR, DENVER CO 80203 (US)

Effective date: 20120801

Owner name: 4350 LA JOLLA VILLAGE DRIVE SUITE 950, SAN DIEGO

Effective date: 20120801

Owner name: AVIGEN, INC.

Effective date: 20120801

Owner name: THE REGENTS OF THE UNIVERSITY OF COLORADO, A BODY

Effective date: 20120801

REG Reference to a national code

Ref country code: PT

Ref legal event code: SC4A

Free format text: AVAILABILITY OF NATIONAL TRANSLATION

Effective date: 20120918

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602008017590

Country of ref document: DE

Effective date: 20120927

Ref country code: DE

Ref legal event code: R082

Ref document number: 602008017590

Country of ref document: DE

Representative=s name: HOFFMANN - EITLE, DE

Ref country code: DE

Ref legal event code: R082

Ref document number: 602008017590

Country of ref document: DE

Representative=s name: HOFFMANN - EITLE PATENT- UND RECHTSANWAELTE PA, DE

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: NOVAGRAAF INTERNATIONAL SA

RAP2 Party data changed (patent owner data changed or rights of a patent transferred)

Owner name: THE REGENTS OF THE UNIVERSITY OF COLORADO, A BODY

Owner name: AVIGEN, INC.

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: NL

Ref legal event code: T3

REG Reference to a national code

Ref country code: NO

Ref legal event code: T2

Effective date: 20120801

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2392116

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20121204

REG Reference to a national code

Ref country code: GR

Ref legal event code: EP

Ref document number: 20120402453

Country of ref document: GR

Effective date: 20121122

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

Effective date: 20120801

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120801

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120801

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121201

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120801

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120801

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120801

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GR

Payment date: 20121228

Year of fee payment: 6

REG Reference to a national code

Ref country code: PL

Ref legal event code: T3

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120801

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120801

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DK

Payment date: 20130110

Year of fee payment: 6

Ref country code: SE

Payment date: 20130114

Year of fee payment: 6

Ref country code: ES

Payment date: 20130207

Year of fee payment: 6

Ref country code: GB

Payment date: 20130123

Year of fee payment: 6

Ref country code: IE

Payment date: 20130110

Year of fee payment: 6

Ref country code: CH

Payment date: 20130114

Year of fee payment: 6

Ref country code: DE

Payment date: 20130123

Year of fee payment: 6

Ref country code: FR

Payment date: 20130204

Year of fee payment: 6

Ref country code: NO

Payment date: 20130109

Year of fee payment: 6

REG Reference to a national code

Ref country code: HK

Ref legal event code: GR

Ref document number: 1139856

Country of ref document: HK

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120801

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20130110

Year of fee payment: 6

Ref country code: BE

Payment date: 20130114

Year of fee payment: 6

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: PT

Payment date: 20130123

Year of fee payment: 6

Ref country code: AT

Payment date: 20121231

Year of fee payment: 6

26N No opposition filed

Effective date: 20130503

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20121101

REG Reference to a national code

Ref country code: HU

Ref legal event code: AG4A

Ref document number: E016466

Country of ref document: HU

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602008017590

Country of ref document: DE

Effective date: 20130503

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130131

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: HU

Payment date: 20130114

Year of fee payment: 6

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130129

Ref country code: CZ

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130129

REG Reference to a national code

Ref country code: PL

Ref legal event code: LAPE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130129

BERE Be: lapsed

Owner name: AVIGEN, INC.

Effective date: 20140131

Owner name: THE REGENTS OF THE UNIVERSITY OF COLORADO, A BODY

Effective date: 20140131

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120801

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602008017590

Country of ref document: DE

REG Reference to a national code

Ref country code: PT

Ref legal event code: MM4A

Free format text: LAPSE DUE TO NON-PAYMENT OF FEES

Effective date: 20140729

REG Reference to a national code

Ref country code: NL

Ref legal event code: V1

Effective date: 20140801

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20140131

REG Reference to a national code

Ref country code: AT

Ref legal event code: MM01

Ref document number: 568267

Country of ref document: AT

Kind code of ref document: T

Effective date: 20140129

REG Reference to a national code

Ref country code: SE

Ref legal event code: EUG

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20140129

REG Reference to a national code

Ref country code: GR

Ref legal event code: ML

Ref document number: 20120402453

Country of ref document: GR

Effective date: 20140801

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602008017590

Country of ref document: DE

Effective date: 20140801

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140131

Ref country code: GR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140801

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140801

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140131

Ref country code: NL

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140801

Ref country code: NO

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140131

REG Reference to a national code

Ref country code: FR

Ref legal event code: ST

Effective date: 20140930

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140131

Ref country code: AT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140129

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140129

Ref country code: HU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140130

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140130

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140729

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140131

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140129

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140131

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20120801

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20130129

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140129

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: ES

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20140130